1
|
Li Z, Ren K, Chen J, Zhuang Y, Dong S, Wang J, Liu H, Ding J. Bioactive hydrogel formulations for regeneration of pathological bone defects. J Control Release 2025; 380:686-714. [PMID: 39880040 DOI: 10.1016/j.jconrel.2025.01.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 01/18/2025] [Accepted: 01/21/2025] [Indexed: 01/31/2025]
Abstract
Bone defects caused by osteoporosis, infection, diabetes, post-tumor resection, and nonunion often cause severe pain and markedly increase morbidity and mortality, which remain a significant challenge for orthopedic surgeons. The precise local treatments for these pathological complications are essential to avoid poor or failed bone repair. Hydrogel formulations serve as injectable innovative platforms that overcome microenvironmental obstacles and as delivery systems for controlled release of various bioactive substances to bone defects in a targeted manner. Additionally, hydrogel formulations can be tailored for specific mechanical strengths and degradation profiles by adjusting their physical and chemical properties, which are crucial for prolonged drug retention and effective bone repair. This review summarizes recent advances in bioactive hydrogel formulations as three-dimensional scaffolds that support cell proliferation and differentiation. It also highlights their role as smart drug-delivery systems with capable of continuously releasing antibacterial agents, anti-inflammatory drugs, chemotherapeutic agents, and osteogenesis-related factors to enhance bone regeneration in pathological areas. Furthermore, the limitations of hydrogel formulations in pathological bone repair are discussed, and future development directions are proposed, which is expected to pave the way for the repair of pathological bone defects.
Collapse
Affiliation(s)
- Zuhao Li
- Orthopaedic Medical Center, The Second Hospital of Jilin University, 4026 Yatai Street, Changchun 130041, China
| | - Kaixuan Ren
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China; Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, 333 Nanchen Road, Shanghai 200444, China
| | - Jiajia Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China; The First Outpatient Department, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 6822 Jinhu Road, Changchun 130021, China
| | - Yaling Zhuang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China
| | - Shujun Dong
- The First Outpatient Department, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 6822 Jinhu Road, Changchun 130021, China
| | - Jincheng Wang
- Orthopaedic Medical Center, The Second Hospital of Jilin University, 4026 Yatai Street, Changchun 130041, China
| | - He Liu
- Orthopaedic Medical Center, The Second Hospital of Jilin University, 4026 Yatai Street, Changchun 130041, China.
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China
| |
Collapse
|
2
|
Li Y, Li W, Li L, Yan C, Wang X, Xiang C, Jia L, Li Q, Zhong X, Jiang K, Chen L. Treating critical bone defects by using core-shell biological scaffold to regulate Fibrosis-Osteogenic homeostasis. Mater Today Bio 2025; 31:101560. [PMID: 40083837 PMCID: PMC11904517 DOI: 10.1016/j.mtbio.2025.101560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 01/12/2025] [Accepted: 02/05/2025] [Indexed: 03/16/2025] Open
Abstract
Critical bone defects pose a significant challenge in the realm of bone defect repair. During the repair process, bone formation is crucial, as the occurrence of invasive tissue growth into the defect, known as fibrosis, is also a possibility. Excessive fibrosis can lead to a "filling effect," wherein fibrous tissue occupies the bone defect area, thereby impeding the bone formation and repair processes. Hence, regulating the dynamic balance between fibrosis and osteogenesis is pivotal to effectively treat critical bone defects. To mitigate the rapid fibrosis rate at the bone defect site, which may result in repair failure, we have devised and fabricated a biomimetic core-shell scaffold-PCL-FAPI/GelMA/HAMA-GBA@plasmid-knockdown SHN-3 (PCL-FAPI/GH-GBA@pk SHN-3)-aimed at modulating fibrosis and vascularization processes within the new callus. The outer "shell" structure of the scaffold employs polycaprolactone (PCL) electrospun nanofibers loaded with fibroblast activating protein inhibitor (FAPI). Utilizing hydrophobic PCL electrospun fibers effectively impedes the growth of exogenous fibrous tissue, while releasing FAPI to inhibit the growth of endogenous fibroblasts. The inner layer "nucleus" structure comprises GelMA/HAMA hydrogel-supported plasmid/polyamideamine (GBA@plasmid-knockdown SHN-3), which enhances the secretion of Slit3 protein and promotes the formation of Type H blood vessels by silencing the SHN-3 gene in osteoblasts. The biomimetic "core-shell" scaffold PCL-FAPI/GH-GBA@pkSHN-3 serves to prevent excessive fibrosis of the callus and foster the formation of Type H blood vessels within the new callus, effectively averting bone nonunion and expediting the repair process of critical bone defects.
Collapse
Affiliation(s)
- Yonghang Li
- Department of Orthopedics, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China
- Department of Joint Orthopedics, Affiliated Hospital of JiangSu University, Zhenjiang, 212000, China
| | - Wenming Li
- Department of Orthopedics, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China
| | - Linfeng Li
- Department of Orthopedics, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China
| | - Caiping Yan
- Department of Orthopedics, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China
| | - Xingkuan Wang
- Department of Orthopedics, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China
| | - Chao Xiang
- Department of Orthopedics, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China
| | - Lifu Jia
- Department of Orthopedics, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China
| | - Qinsong Li
- Department of Orthopedics, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China
| | - Xuemei Zhong
- School of Clinical Medicine, Chongqing Medical and Pharmaceutical College, No. 82, Daxuecheng Zhong Rd, Shapingba Dist, Chongqing, 401331, China
| | - Ke Jiang
- Department of Orthopedics, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China
| | - Lu Chen
- Department of Orthopedics, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China
| |
Collapse
|
3
|
Wu J, Li R, Liu C, Li W. The role of AKR1B10 in osteogenic differentiation of mesenchymal stem cells and atrophic nonunion. Bone 2024; 190:117284. [PMID: 39401534 DOI: 10.1016/j.bone.2024.117284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/29/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024]
Abstract
Atrophic nonunion is a chronic disease without effective medications. Here, high-throughput mRNA sequencing was used to explore the novel targets in atrophic nonunion. AKR1B10, a member of aldo-keto reductase family 1, is upregulated in atrophic nonunion tissues. There are currently no studies to reveal the role of AKR1B10 in atrophic nonunion. We used rat bone marrow-derived mesenchymal stem cells (BMSCs) to explore the effect of AKR1B10 on the osteogenic differentiation and autophagy. In vivo, we implanted collagen sponges loaded with LV-shAKR1B10-transduced BMSCs into rat fractured femurs to explore the role of AKR1B10 in fracture healing. The results showed that AKR1B10 reduced the activity of ALP, suppressed the expression of COL1A1, RUNX2 and OCN, and inhibited calcification deposition in osteogenically differentiated BMSCs. AKR1B10 reduced the expression of LC3II, decreased the number of autophagosomes, and promoted the expression of p62. In addition, the promoting effect of AKR1B10 knockdown on osteogenic differentiation of BMSCs was attenuated by 3-MA treatment. Implantation of collagen sponges found that knockdown of AKR1B10 promoted bone fracture healing. In conclusion, AKR1B10 inhibited the osteogenic differentiation and autophagy, and delayed the bone fracture healing. These results provide a new perspective on revealing the role of AKR1B10 in nonunion and may also provide a new therapeutic target for the treatment of nonunion.
Collapse
Affiliation(s)
- Jie Wu
- Department of Orthopaedics, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Runze Li
- Department of Orthopaedics, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Chen Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Weiming Li
- Department of Orthopaedics, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China.
| |
Collapse
|
4
|
Liu YL, Tang XT, Shu HS, Zou W, Zhou BO. Fibrous periosteum repairs bone fracture and maintains the healed bone throughout mouse adulthood. Dev Cell 2024; 59:1192-1209.e6. [PMID: 38554700 DOI: 10.1016/j.devcel.2024.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 12/07/2023] [Accepted: 03/06/2024] [Indexed: 04/02/2024]
Abstract
Bone is regarded as one of few tissues that heals without fibrous scar. The outer layer of the periosteum is covered with fibrous tissue, whose function in bone formation is unknown. We herein developed a system to distinguish the fate of fibrous-layer periosteal cells (FL-PCs) from the skeletal stem/progenitor cells (SSPCs) in the cambium-layer periosteum and bone marrow in mice. We showed that FL-PCs did not participate in steady-state osteogenesis, but formed the main body of fibrocartilaginous callus during fracture healing. Moreover, FL-PCs invaded the cambium-layer periosteum and bone marrow after fracture, forming neo-SSPCs that continued to maintain the healed bones throughout adulthood. The FL-PC-derived neo-SSPCs expressed lower levels of osteogenic signature genes and displayed lower osteogenic differentiation activity than the preexisting SSPCs. Consistent with this, healed bones were thinner and formed more slowly than normal bones. Thus, the fibrous periosteum becomes the cellular origin of bones after fracture and alters bone properties permanently.
Collapse
Affiliation(s)
- Yiming Liam Liu
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Xinyu Thomas Tang
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Hui Sophie Shu
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Weiguo Zou
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; Key Laboratory of RNA Science and Engineering, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.
| | - Bo O Zhou
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences, Tianjin 300020, China.
| |
Collapse
|
5
|
Zhang X, Deng C, Qi S. Periosteum Containing Implicit Stem Cells: A Progressive Source of Inspiration for Bone Tissue Regeneration. Int J Mol Sci 2024; 25:2162. [PMID: 38396834 PMCID: PMC10889827 DOI: 10.3390/ijms25042162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/12/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
The periosteum is known as the thin connective tissue covering most bone surfaces. Its extrusive bone regeneration capacity was confirmed from the very first century-old studies. Recently, pluripotent stem cells in the periosteum with unique physiological properties were unveiled. Existing in dynamic contexts and regulated by complex molecular networks, periosteal stem cells emerge as having strong capabilities of proliferation and multipotential differentiation. Through continuous exploration of studies, we are now starting to acquire more insight into the great potential of the periosteum in bone formation and repair in situ or ectopically. It is undeniable that the periosteum is developing further into a more promising strategy to be harnessed in bone tissue regeneration. Here, we summarized the development and structure of the periosteum, cell markers, and the biological features of periosteal stem cells. Then, we reviewed their pivotal role in bone repair and the underlying molecular regulation. The understanding of periosteum-related cellular and molecular content will help enhance future research efforts and application transformation of the periosteum.
Collapse
Affiliation(s)
- Xinyuan Zhang
- Department of Prosthodontics, Shanghai Stomatological Hospital, School of Stomatology, Fudan University, Shanghai 200001, China;
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai 200001, China
| | - Chen Deng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China;
| | - Shengcai Qi
- Department of Prosthodontics, Shanghai Stomatological Hospital, School of Stomatology, Fudan University, Shanghai 200001, China;
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai 200001, China
| |
Collapse
|
6
|
Cao Y, Bolam SM, Boss AL, Murray HC, Munro JT, Poulsen RC, Dalbeth N, Brooks AES, Matthews BG. Characterization of adult human skeletal cells in different tissues reveals a CD90 +CD34 + periosteal stem/progenitor population. Bone 2024; 178:116926. [PMID: 37793499 DOI: 10.1016/j.bone.2023.116926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/27/2023] [Accepted: 10/01/2023] [Indexed: 10/06/2023]
Abstract
The periosteum plays a crucial role in bone healing and is an important source of skeletal stem and progenitor cells. Recent studies in mice indicate that diverse populations of skeletal progenitors contribute to growth, homeostasis and healing. Information about the in vivo identity and diversity of skeletal stem and progenitor cells in different compartments of the adult human skeleton is limited. In this study, we compared non-hematopoietic populations in matched tissues from the femoral head and neck of 21 human participants using spectral flow cytometry of freshly isolated cells. High-dimensional clustering analysis indicated significant differences in marker distribution between periosteum, articular cartilage, endosteum and bone marrow populations, and identified populations that were highly enriched or unique to specific tissues. Periosteum-enriched markers included CD90 and CD34. Articular cartilage, which has very poor regenerative potential, showed enrichment of multiple markers, including the PDPN+CD73+CD164+CD146- population previously reported to represent human skeletal stem cells. We further characterized periosteal populations by combining CD90 with other strongly expressed markers. CD90+CD34+ cells sorted directly from periosteum showed significant colony-forming unit fibroblasts (CFU-F) enrichment, rapid expansion, and consistent multi-lineage differentiation of clonal populations in vitro. In situ, CD90+CD34+ cells include a perivascular population in the outer layer of the periosteum and non-perivascular cells closer to the bone surface. CD90+ cells are also highly enriched for CFU-F in bone marrow and endosteum, but not articular cartilage. In conclusion, our study indicates considerable diversity in the non-hematopoietic cell populations in different tissue compartments within the adult human skeleton, and suggests that periosteal progenitor cells reside within the CD90+CD34+ population.
Collapse
Affiliation(s)
- Ye Cao
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Scott M Bolam
- Department of Surgery, University of Auckland, Auckland, New Zealand
| | - Anna L Boss
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
| | - Helen C Murray
- Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Jacob T Munro
- Department of Surgery, University of Auckland, Auckland, New Zealand
| | - Raewyn C Poulsen
- Department of Pharmacology, University of Auckland, Auckland, New Zealand
| | - Nicola Dalbeth
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Anna E S Brooks
- School of Biological Sciences, University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Brya G Matthews
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
7
|
Voss JO, Bolis R, Koerdt S, Doll C, Rubarth K, Duda GN, Heiland M, Fischer H, Rendenbach C, Ebker T, Steffen C. Quantifying bone healing after mandibular displacement in orthognathic surgery. Br J Oral Maxillofac Surg 2024; 62:45-50. [PMID: 38008679 DOI: 10.1016/j.bjoms.2023.10.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/13/2023] [Indexed: 11/28/2023]
Abstract
Impaired bony healing following bilateral sagittal split osteotomy (BSSO) is a major unmet medical need for affected patients, and rare occurrences can hinder the identification of underlying risk factors. We hypothesised that osseous union following BSSO can be quantified using volumetric analysis, and we aimed to identify the risk factors for impaired bone healing. The percentage change in bony volume was measured in orthognathic patients following BSSO using two consecutive postoperative cone-beam computed tomography scans. Patients' characteristics and treatment parameters were documented, and correlation and regression analyses of these variables performed. Thirty-six patients (23 men and 13 women) with a mean (SD) age of 33.28 (11.86) years were included. The gap site (lingual versus buccal) (p < 0.01) had a significant impact on the change in volume. Age (p = 0.06) showed a trend towards significance. Initial width of the osteotomy gap, sex, and indication for surgery did not influence osseous healing. Increased age at surgery and the side of the buccal osteotomy are independent risk factors for impaired osseous healing following BSSO.
Collapse
Affiliation(s)
- Jan Oliver Voss
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Oral and Maxillofacial Surgery, Augustenburger Platz 1, 13353 Berlin, Germany; Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Straße 2, 10178 Berlin, Germany.
| | - Rwis Bolis
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Oral and Maxillofacial Surgery, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Steffen Koerdt
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Oral and Maxillofacial Surgery, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Christian Doll
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Oral and Maxillofacial Surgery, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Kerstin Rubarth
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Biometry and Clinical Epidemiology, Charitéplatz 1, 10117 Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Informatics, Charitéplatz 1, 10117 Berlin, Germany
| | - Georg N Duda
- Julius Wolff Institute and Berlin Institute of Health Centre for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Straße 2, 10178 Berlin, Germany; Charité - Universitätsmedizin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Center for Musculoskeletal Surgery, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Max Heiland
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Oral and Maxillofacial Surgery, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Heilwig Fischer
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Oral and Maxillofacial Surgery, Augustenburger Platz 1, 13353 Berlin, Germany; Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Straße 2, 10178 Berlin, Germany; Charité - Universitätsmedizin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Center for Musculoskeletal Surgery, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Carsten Rendenbach
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Oral and Maxillofacial Surgery, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Tobias Ebker
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Oral and Maxillofacial Surgery, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Claudius Steffen
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Oral and Maxillofacial Surgery, Augustenburger Platz 1, 13353 Berlin, Germany
| |
Collapse
|
8
|
Yao L, Lu J, Zhong L, Wei Y, Gui T, Wang L, Ahn J, Boerckel JD, Rux D, Mundy C, Qin L, Pacifici M. Activin A marks a novel progenitor cell population during fracture healing and reveals a therapeutic strategy. eLife 2023; 12:e89822. [PMID: 38079220 PMCID: PMC10783872 DOI: 10.7554/elife.89822] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 12/08/2023] [Indexed: 12/18/2023] Open
Abstract
Insufficient bone fracture repair represents a major clinical and societal burden and novel strategies are needed to address it. Our data reveal that the transforming growth factor-β superfamily member Activin A became very abundant during mouse and human bone fracture healing but was minimally detectable in intact bones. Single-cell RNA-sequencing revealed that the Activin A-encoding gene Inhba was highly expressed in a unique, highly proliferative progenitor cell (PPC) population with a myofibroblast character that quickly emerged after fracture and represented the center of a developmental trajectory bifurcation producing cartilage and bone cells within callus. Systemic administration of neutralizing Activin A antibody inhibited bone healing. In contrast, a single recombinant Activin A implantation at fracture site in young and aged mice boosted: PPC numbers; phosphorylated SMAD2 signaling levels; and bone repair and mechanical properties in endochondral and intramembranous healing models. Activin A directly stimulated myofibroblastic differentiation, chondrogenesis and osteogenesis in periosteal mesenchymal progenitor culture. Our data identify a distinct population of Activin A-expressing PPCs central to fracture healing and establish Activin A as a potential new therapeutic tool.
Collapse
Affiliation(s)
- Lutian Yao
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Department of Orthopaedics, The First Hospital of China Medical UniversityShenyangChina
| | - Jiawei Lu
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Leilei Zhong
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Yulong Wei
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Tao Gui
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Luqiang Wang
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Jaimo Ahn
- Department of Orthopaedic Surgery, Michigan Medicine, University of MichiganAnn ArborUnited States
| | - Joel D Boerckel
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Danielle Rux
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Christina Mundy
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Ling Qin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Maurizio Pacifici
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
| |
Collapse
|
9
|
Mundy C, Yao L, Shaughnessy KA, Saunders C, Shore EM, Koyama E, Pacifici M. Palovarotene Action Against Heterotopic Ossification Includes a Reduction of Local Participating Activin A-Expressing Cell Populations. JBMR Plus 2023; 7:e10821. [PMID: 38130748 PMCID: PMC10731142 DOI: 10.1002/jbm4.10821] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 09/09/2023] [Indexed: 12/23/2023] Open
Abstract
Heterotopic ossification (HO) consists of extraskeletal bone formation. One form of HO is acquired and instigated by traumas or surgery, and another form is genetic and characterizes fibrodysplasia ossificans progressiva (FOP). Recently, we and others showed that activin A promotes both acquired and genetic HO, and in previous studies we found that the retinoid agonist palovarotene inhibits both HO forms in mice. Here, we asked whether palovarotene's action against HO may include an interference with endogenous activin A expression and/or function. Using a standard mouse model of acquired HO, we found that activin A and its encoding RNA (Inhba) were prominent in chondrogenic cells within developing HO masses in untreated mice. Single-cell RNAseq (scRNAseq) assays verified that Inhba expression characterized chondroprogenitors and chondrocytes in untreated HO, in addition to its expected expression in inflammatory cells and macrophages. Palovarotene administration (4 mg/kg/d/gavage) caused a sharp inhibition of both HO and amounts of activin A and Inhba transcripts. Bioinformatic analyses of scRNAseq data sets indicated that the drug had reduced interactions and cross-talk among local cell populations. To determine if palovarotene inhibited Inhba expression directly, we assayed primary chondrocyte cultures. Drug treatment inhibited their cartilaginous phenotype but not Inhba expression. Our data reveal that palovarotene markedly reduces the number of local Inhba-expressing HO-forming cell populations. The data broaden the spectrum of HO culprits against which palovarotene acts, accounting for its therapeutic effectiveness. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Christina Mundy
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic SurgeryThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
| | - Lutian Yao
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic SurgeryThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
- Department of OrthopaedicsThe First Hospital of China Medical UniversityShenyangChina
| | - Kelly A. Shaughnessy
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic SurgeryThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
| | - Cheri Saunders
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic SurgeryThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
| | - Eileen M. Shore
- Departments of Orthopaedic Surgery and Genetics, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Eiki Koyama
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic SurgeryThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
| | - Maurizio Pacifici
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic SurgeryThe Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
| |
Collapse
|
10
|
Moore ER, Maridas DE, Gamer L, Chen G, Burton K, Rosen V. A periosteum-derived cell line to study the role of BMP/TGFβ signaling in periosteal cell behavior and function. Front Physiol 2023; 14:1221152. [PMID: 37799511 PMCID: PMC10547901 DOI: 10.3389/fphys.2023.1221152] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 09/05/2023] [Indexed: 10/07/2023] Open
Abstract
The periosteum is a thin tissue surrounding each skeletal element that contains stem and progenitor cells involved in bone development, postnatal appositional bone growth, load-induced bone formation, and fracture repair. BMP and TGFβ signaling are important for periosteal activity and periosteal cell behavior, but thorough examination of the influence of these pathways on specific cell populations resident in the periosteum is lacking due to limitations associated with primary periosteal cell isolations and in vitro experiments. Here we describe the generation of a novel periosteum-derived clonal cell (PDC) line from postnatal day 14 mice and use it to examine periosteal cell behavior in vitro. PDCs exhibit key characteristics of periosteal cells observed during skeletal development, maintenance, and bone repair. Specifically, PDCs express established periosteal markers, can be expanded in culture, demonstrate the ability to differentiate into chondrocytes, osteoblasts, and adipocytes, and exhibit an osteogenic response to physical stimulation. PDCs also engage in BMP and/or TGFβ signaling when treated with the activating ligands BMP2 and TGFβ-1, and in response to mechanical stimulation via fluid shear. We believe that this PDC line will be useful for large-scale, long-term experiments that were not feasible when using primary periosteal cells. Anticipated future uses include advancing our understanding of the signaling interactions that occur during appositional bone growth and fracture repair and developing drug screening platforms to discover novel growth and fracture healing factors.
Collapse
Affiliation(s)
- Emily R. Moore
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA, United States
| | | | | | | | | | | |
Collapse
|
11
|
Hu M, Zeng W, Zhang J, Feng Y, Ma L, Huang F, Cai Q. Fixators dynamization for delayed union and non-union of femur and tibial fractures: a review of techniques, timing and influence factors. J Orthop Surg Res 2023; 18:577. [PMID: 37550732 PMCID: PMC10405409 DOI: 10.1186/s13018-023-04054-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/27/2023] [Indexed: 08/09/2023] Open
Abstract
The optimal balance between mechanical environment and biological factors is crucial for successful bone healing, as they synergistically affect bone development. Any imbalance between these factors can lead to impaired bone healing, resulting in delayed union or non-union. To address this bone healing disorder, clinicians have adopted a technique known as "dynamization" which involves modifying the stiffness properties of the fixator. This technique facilitates the establishment of a favorable mechanical and biological environment by changing a rigid fixator to a more flexible one that promotes bone healing. However, the dynamization of fixators is selective for certain types of non-union and can result in complications or failure to heal if applied to inappropriate non-unions. This review aims to summarize the indications for dynamization, as well as introduce a novel dynamic locking plate and various techniques for dynamization of fixators (intramedullary nails, steel plates, external fixators) in femur and tibial fractures. Additionally, Factors associated with the effectiveness of dynamization are explored in response to the variation in dynamization success rates seen in clinical studies.
Collapse
Affiliation(s)
- Minhua Hu
- The First Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenxing Zeng
- The First Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jingtao Zhang
- The First Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuanlan Feng
- The First Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Luyao Ma
- The First Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Feng Huang
- The First Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China.
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Qunbin Cai
- The First Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China.
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
12
|
Marcucio R, Miclau T, Bahney C. A Shifting Paradigm: Transformation of Cartilage to Bone during Bone Repair. J Dent Res 2023; 102:13-20. [PMID: 36303415 PMCID: PMC9791286 DOI: 10.1177/00220345221125401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
While formation and regeneration of the skeleton have been studied for a long period of time, significant scientific advances in this field continue to emerge based on an unmet clinical need to improve options to promote bone repair. In this review, we discuss the relationship between mechanisms of bone formation and bone regeneration. Data clearly show that regeneration is not simply a reinduction of the molecular and cellular programs that were used for development. Instead, the mechanical environment exerts a strong influence on the mode of repair, while during development, cell-intrinsic processes drive the mode of skeletal formation. A major advance in the field has shown that cell fate is flexible, rather than terminal, and that chondrocytes are able to differentiate into osteoblasts and other cell types during development and regeneration. This is discussed in a larger context of regeneration in vertebrates as well as the clinical implication that this shift in understanding presents.
Collapse
Affiliation(s)
- R.S. Marcucio
- University of California, San Francisco (UCSF), Orthopaedic Trauma Institute, San Francisco, CA, USA
| | - T. Miclau
- University of California, San Francisco (UCSF), Orthopaedic Trauma Institute, San Francisco, CA, USA
| | - C.S. Bahney
- University of California, San Francisco (UCSF), Orthopaedic Trauma Institute, San Francisco, CA, USA
- Steadman Philippon Research Institute, Vail, CO, USA
| |
Collapse
|
13
|
Hixon KR, Katz DB, McKenzie JA, Miller AN, Guilak F, Silva MJ. Cryogel Scaffold-Mediated Delivery of Adipose-Derived Stem Cells Promotes Healing in Murine Model of Atrophic Non-Union. Front Bioeng Biotechnol 2022; 10:851904. [PMID: 35600896 PMCID: PMC9117654 DOI: 10.3389/fbioe.2022.851904] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/23/2022] [Indexed: 01/08/2023] Open
Abstract
Non-union is defined as the permanent failure of a bone to heal and occurs clinically in 5% of fractures. Atrophic non-unions, characterized by absent/minimal callus formation, are poorly understood and difficult to treat. We recently demonstrated a novel murine model of atrophic non-union in the 3.6Col1A1-tk (Col1-tk) mouse, wherein dosing with the nucleoside analog ganciclovir (GCV) was used to deplete proliferating osteoprogenitor cells, leading to a radiographic and biomechanical non-union after the mid-shaft femur fracture. Using this Col1-tk atrophic non-union model, we hypothesized that the scaffold-mediated lentiviral delivery of doxycycline-inducible BMP-2 transgenes would induce osteogenesis at the fracture site. Cryogel scaffolds were used as a vehicle for GFP+ and BMP-2+ cell delivery to the site of non-union. Cryogel scaffolds were biofabricated through the cross-linking of a chitosan-gelatin polymer solution at subzero temperatures, which results in a macroporous, spongy structure that may be advantageous for a bone regeneration application. Murine adipose-derived stem cells were seeded onto the cryogel scaffolds, where they underwent lentiviral transduction. Following the establishment of atrophic non-unions in the femurs of Col1-tk mice (4 weeks post-fracture), transduced, seeded scaffolds were surgically placed around the site of non-union, and the animals were given doxycycline water to induce BMP-2 production. Controls included GFP+ cells on the cryogel scaffolds, acellular scaffolds, and sham (no scaffold). Weekly radiographs were taken, and endpoint analysis included micro-CT and histological staining. After 2 weeks of implantation, the BMP-2+ scaffolds were infiltrated with cartilage and woven bone at the non-union site, while GFP+ scaffolds had woven bone formation. Later, timepoints of 8 weeks had woven bone and vessel formation within the BMP-2+ and GFP + scaffolds with cortical bridging of the original fracture site in both groups. Overall, the cell-seeded cryogels promoted osseous healing. However, while the addition of BMP-2 promoted the endochondral ossification, it may provide a slower route to healing. This proof-of-concept study demonstrates the potential for cellularized cryogel scaffolds to enhance the healing of non-unions.
Collapse
Affiliation(s)
- Katherine R. Hixon
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University, St. Louis, MO, United States
- Thayer School of Engineering, Dartmouth College, Hanover, NH, United States
| | - Dakota B. Katz
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University, St. Louis, MO, United States
- Department of Biomedical Engineering, Washington University, St. Louis, MO, United States
- Center of Regenerative Medicine, Washington University, St. Louis, MO, United States
- Shriners Hospitals for Children—St. Louis, St. Louis, MO, United States
| | - Jennifer A. McKenzie
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University, St. Louis, MO, United States
| | - Anna N. Miller
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University, St. Louis, MO, United States
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University, St. Louis, MO, United States
- Department of Biomedical Engineering, Washington University, St. Louis, MO, United States
- Center of Regenerative Medicine, Washington University, St. Louis, MO, United States
- Shriners Hospitals for Children—St. Louis, St. Louis, MO, United States
| | - Matthew J. Silva
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University, St. Louis, MO, United States
- Department of Biomedical Engineering, Washington University, St. Louis, MO, United States
- Center of Regenerative Medicine, Washington University, St. Louis, MO, United States
| |
Collapse
|
14
|
Hu Y, He Y, Fang J, Liu Y, Cao Y, Tong W, Chen W, Shao Z, Liu Y, Tian H. Wnt10b-overexpressing umbilical cord mesenchymal stem cells promote fracture healing via accelerated cartilage callus to bone remodeling. Bioengineered 2022; 13:10313-10323. [PMID: 35436412 PMCID: PMC9161882 DOI: 10.1080/21655979.2022.2062954] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
The aim of this study was to investigate whether HUCMSCsWnt10b could promote long bone fracture healing. Commercially-available HUCMSCsEmp (human umbilical cord mesenchymal stem cells transfected with empty vector) in hydrogel, HUCMSCsWnt10b in hydrogel and HUCMSCsWnt10b with the Wnt signaling pathway inhibitor IWR-1 were transplanted into the fracture site in a rat model of femoral fracture. We found that transplantation of HUCMSCsWnt10b significantly accelerated bone healing in a rat model of femoral fracture. Meanwhile, three-point bending test proved that the mechanical properties of the bone at the fracture site in the HUCMSCWnt10b treatment group were significantly better than those of the other treatment groups. To understand the cellular mechanism, we explored the viability of periosteal stem cells (PSCs), as they contribute the greatest number of osteoblast lineage cells to the callus. In line with in vivo data, we found that conditioned medium from HUCMSCsWnt10b enhanced the migration and osteogenic differentiation of PSCs. Furthermore, conditioned medium from HUCMSCsWnt10b also induced endothelial cells to form capillary-like structures in a tube formation assay, which was blocked by SU5416, an angiogenesis inhibitor, suggesting that enhanced vessel formation and growth also contribute to accelerated hard callus formation. In summary, our study demonstrates that HUCMSCsWnt10b promote fracture healing via accelerated hard callus formation, possibly due to enhanced osteogenic differentiation of PSCs and vessel growth. Therefore, HUCMSCsWnt10b may be a promising treatment for long bone fractures.
Collapse
Affiliation(s)
- Yuxiang Hu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Hubei, China
| | - Yu He
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Hubei, China
| | - Jiarui Fang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Hubei, China
| | - Yunlu Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Hubei, China
| | - Yulin Cao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Hubei, China
| | - Wei Tong
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Hubei, China
| | - Wei Chen
- Department of Orthopedics, The Third Hospital of Hebei Medical University, Shi Jiazhuang, Hebei, China.,Nhc Key Laboratory of Intelligent Orthopedic Equipment (The Third Hospital of Hebei Medical University), Shi Jiazhuang, Hebei, China
| | - Zengwu Shao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Hubei, China
| | - Yong Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Hubei, China
| | - Hongtao Tian
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Hubei, China
| |
Collapse
|
15
|
Hixon KR, Miller AN. Animal models of impaired long bone healing and tissue engineering- and cell-based in vivo interventions. J Orthop Res 2022; 40:767-778. [PMID: 35072292 DOI: 10.1002/jor.25277] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 12/05/2021] [Accepted: 01/16/2022] [Indexed: 02/04/2023]
Abstract
Bone healing after injury typically follows a systematic process and occurs spontaneously under appropriate physiological conditions. However, impaired long bone healing is still quite common and may require surgical intervention. Various complications can result in different forms of impaired bone healing including nonunion, critical-size defects, or stress fractures. While a nonunion may occur due to impaired biological signaling and/or mechanical instability, a critical-size defect exhibits extensive bone loss that will not spontaneously heal. Comparatively, a stress fracture occurs from repetitive forces and results in a non-healing crack or break in the bone. Clinical standards of treatment vary between these bone defects due to their pathological differences. The use of appropriate animal models for modeling healing defects is critical to improve current treatment methods and develop novel rescue therapies. This review provides an overview of these clinical bone healing impairments and current animal models available to study the defects in vivo. The techniques used to create these models are compared, along with the outcomes, to clarify limitations and future objectives. Finally, rescue techniques focused on tissue engineering and cell-based therapies currently applied in animal models are specifically discussed to analyze their ability to initiate healing at the defect site, providing information regarding potential future therapies. In summary, this review focuses on the current animal models of nonunion, critical-size defects, and stress fractures, as well as interventions that have been tested in vivo to provide an overview of the clinical potential and future directions for improving bone healing.
Collapse
Affiliation(s)
- Katherine R Hixon
- Department of Orthopaedic Surgery, Washington University, St. Louis, Missouri, USA.,Thayer School of Engineering, Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| | - Anna N Miller
- Department of Orthopaedic Surgery, Washington University, St. Louis, Missouri, USA
| |
Collapse
|
16
|
Cohn-Schwartz D, Schary Y, Yalon E, Krut Z, Da X, Schwarz EM, Gazit D, Pelled G, Gazit Z. PTH-Induced Bone Regeneration and Vascular Modulation Are Both Dependent on Endothelial Signaling. Cells 2022; 11:cells11050897. [PMID: 35269519 PMCID: PMC8909576 DOI: 10.3390/cells11050897] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 02/28/2022] [Accepted: 03/02/2022] [Indexed: 12/10/2022] Open
Abstract
The use of a bone allograft presents a promising approach for healing nonunion fractures. We have previously reported that parathyroid hormone (PTH) therapy induced allograft integration while modulating angiogenesis at the allograft proximity. Here, we hypothesize that PTH-induced vascular modulation and the osteogenic effect of PTH are both dependent on endothelial PTH receptor-1 (PTHR1) signaling. To evaluate our hypothesis, we used multiple transgenic mouse lines, and their wild-type counterparts as a control. In addition to endothelial-specific PTHR1 knock-out mice, we used mice in which PTHR1 was engineered to be constitutively active in collagen-1α+ osteoblasts, to assess the effect of PTH signaling activation exclusively in osteoprogenitors. To characterize resident cell recruitment and osteogenic activity, mice in which the Luciferase reporter gene is expressed under the Osteocalcin promoter (Oc-Luc) were used. Mice were implanted with calvarial allografts and treated with either PTH or PBS. A micro-computed tomography-based structural analysis indicated that the induction of bone formation by PTH, as observed in wild-type animals, was not maintained when PTHR1 was removed from endothelial cells. Furthermore, the induction of PTH signaling exclusively in osteoblasts resulted in significantly less bone formation compared to systemic PTH treatment, and significantly less osteogenic activity was measured by bioluminescence imaging of the Oc-Luc mice. Deletion of the endothelial PTHR1 significantly decreased the PTH-induced formation of narrow blood vessels, formerly demonstrated in wild-type mice. However, the exclusive activation of PTH signaling in osteoblasts was sufficient to re-establish the observed PTH effect. Collectively, our results show that endothelial PTHR1 signaling plays a key role in PTH-induced osteogenesis and has implications in angiogenesis.
Collapse
Affiliation(s)
- Doron Cohn-Schwartz
- Department of Internal Medicine B, Division of Internal Medicine, Rambam Healthcare Campus, Haifa 3109601, Israel;
- Skeletal Biotech Laboratory, Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel; (Y.S.); (E.Y.); (D.G.); (G.P.)
| | - Yeshai Schary
- Skeletal Biotech Laboratory, Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel; (Y.S.); (E.Y.); (D.G.); (G.P.)
| | - Eran Yalon
- Skeletal Biotech Laboratory, Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel; (Y.S.); (E.Y.); (D.G.); (G.P.)
| | - Zoe Krut
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Xiaoyu Da
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
| | - Edward M. Schwarz
- The Center for Musculoskeletal Research, Department of Orthopaedics, School of Medicine & Dentistry, University of Rochester, Rochester, NY 14642, USA;
| | - Dan Gazit
- Skeletal Biotech Laboratory, Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel; (Y.S.); (E.Y.); (D.G.); (G.P.)
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
- Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Gadi Pelled
- Skeletal Biotech Laboratory, Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel; (Y.S.); (E.Y.); (D.G.); (G.P.)
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
- Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Zulma Gazit
- Skeletal Biotech Laboratory, Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel; (Y.S.); (E.Y.); (D.G.); (G.P.)
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Correspondence:
| |
Collapse
|
17
|
Hixon KR, McKenzie JA, Sykes DAW, Yoneda S, Hensley A, Buettmann EG, Zheng H, Skouteris D, McAlinden A, Miller AN, Silva MJ. Ablation of Proliferating Osteoblast Lineage Cells After Fracture Leads to Atrophic Nonunion in a Mouse Model. J Bone Miner Res 2021; 36:2243-2257. [PMID: 34405443 PMCID: PMC8719642 DOI: 10.1002/jbmr.4424] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 07/15/2021] [Accepted: 08/10/2021] [Indexed: 01/19/2023]
Abstract
Nonunion is defined as the permanent failure of a fractured bone to heal, often necessitating surgical intervention. Atrophic nonunions are a subtype that are particularly difficult to treat. Animal models of atrophic nonunion are available; however, these require surgical or radiation-induced trauma to disrupt periosteal healing. These methods are invasive and not representative of many clinical nonunions where osseous regeneration has been arrested by a "failure of biology". We hypothesized that arresting osteoblast cell proliferation after fracture would lead to atrophic nonunion in mice. Using mice that express a thymidine kinase (tk) "suicide gene" driven by the 3.6Col1a1 promoter (Col1-tk), proliferating osteoblast lineage cells can be ablated upon exposure to the nucleoside analog ganciclovir (GCV). Wild-type (WT; control) and Col1-tk littermates were subjected to a full femur fracture and intramedullary fixation at 12 weeks age. We confirmed abundant tk+ cells in fracture callus of Col-tk mice dosed with water or GCV, specifically many osteoblasts, osteocytes, and chondrocytes at the cartilage-bone interface. Histologically, we observed altered callus composition in Col1-tk mice at 2 and 3 weeks postfracture, with significantly less bone and more fibrous tissue. Col1-tk mice, monitored for 12 weeks with in vivo radiographs and micro-computed tomography (μCT) scans, had delayed bone bridging and reduced callus size. After euthanasia, ex vivo μCT and histology showed failed union with residual bone fragments and fibrous tissue in Col1-tk mice. Biomechanical testing showed a failure to recover torsional strength in Col1-tk mice, in contrast to WT. Our data indicates that suppression of proliferating osteoblast-lineage cells for at least 2 weeks after fracture blunts the formation and remodeling of a mineralized callus leading to a functional nonunion. We propose this as a new murine model of atrophic nonunion. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Katherine R Hixon
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | - Jennifer A McKenzie
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | - David A W Sykes
- Department of Biology, Washington University in St. Louis, St. Louis, MO, USA
| | - Susumu Yoneda
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | - Austin Hensley
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Evan G Buettmann
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Hongjun Zheng
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | - Dimitrios Skouteris
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | - Audrey McAlinden
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA.,Department of Cell Biology & Physiology, Washington University in St. Louis, St. Louis, MO, USA.,St. Louis Shriners Hospital Research Center, Shriners Hospital for Children, St. Louis, MO, USA
| | - Anna N Miller
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | - Matthew J Silva
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
18
|
Wang L, Yao L, Duan H, Yang F, Lin M, Zhang R, He Z, Ahn J, Fan Y, Qin L, Gong Y. Plasminogen Regulates Fracture Repair by Promoting the Functions of Periosteal Mesenchymal Progenitors. J Bone Miner Res 2021; 36:2229-2242. [PMID: 34378815 PMCID: PMC8865375 DOI: 10.1002/jbmr.4423] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 11/06/2022]
Abstract
Defective or insufficient bone repair and regeneration are common in patients as a result of major trauma or severe disease. Cell therapy with periosteal mesenchymal progenitors, which can be limited in severe injury, serves as a promising approach; however, its efficacy is limited due to a repair-hostile ischemic tissue microenvironment after traumatic fracture. Here we report that plasminogen (Plg), a factor that is upregulated in these environments, is critical for fracture healing. Plg knockout mice had impaired trabecular and cortical bone structure and exhibited delayed and incomplete fracture healing. Interestingly, Plg deficiency greatly reduced the thickness of expanded periosteum, suggesting a role of Plg in periosteal mesenchymal progenitor-mediated bone repair. In culture, Plg increased cell proliferation and migration in periosteal mesenchymal progenitors and inhibited cell death under ischemic conditions. Mechanistically, we revealed that Plg cleaved and activated Cyr61 to regulate periosteal progenitor function. Thus, our study uncovers a cellular mechanism underlying fracture healing, by which Plg activates Cyr61 to promote periosteal progenitor proliferation, survival, and migration and improves bone repair after fracture. Targeting Plg may offer a rational and effective therapeutic opportunity for improving fracture healing. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Luqiang Wang
- Departments of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Orthopaedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lutian Yao
- Departments of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Orthopaedics, The First Hospital of China Medical University, Shenyang, China
| | - Hao Duan
- Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Neurosurgery, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Fan Yang
- Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Maohuan Lin
- Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rongxin Zhang
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhenqiang He
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jaimo Ahn
- Departments of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yi Fan
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ling Qin
- Departments of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yanqing Gong
- Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
19
|
Bai Y, Yu T, Deng J, Yang Y, Tan J, Dai Q, Zhang Z, Dong S, Xu J. Connective Tissue Growth Factor From Periosteal Tartrate Acid Phosphatase-Positive Monocytes Direct Skeletal Stem Cell Renewal and Fate During Bone Healing. Front Cell Dev Biol 2021; 9:730095. [PMID: 34595178 PMCID: PMC8476870 DOI: 10.3389/fcell.2021.730095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 08/20/2021] [Indexed: 11/25/2022] Open
Abstract
The periosteum is critical for bone healing. Studies have shown that the periosteum contains periosteal stem cells (PSCs) with multidirectional differentiation potential and self-renewal ability. PSCs are activated in early fracture healing and are committed to the chondrocyte lineage, which is the basis of callus formation. However, the mechanism by which PSCs are activated and committed to chondrocytes in bone regeneration remains unclear. Here, we show that tartrate acid phosphatase (TRAP)-positive monocytes secrete CTGF to activate PSCs during bone regeneration. The loss function of TRAP-positive monocytes identifies their specific role during bone healing. Then, the secreted CTGF promotes endochondral ossification and activates PSCs in mouse bone fracture models. The secreted CTGF enhances PSC renewal by upregulating the expression of multiple pluripotent genes. CTGF upregulates c-Jun expression through αVβ5 integrin. Then, c-Jun transcription activates the transcription of the pluripotent genes Sox2, Oct4, and Nanog. Simultaneously, CTGF also activates the transcription and phosphorylation of Smad3 through αVβ5 integrin, which is the central gene in chondrogenesis. Our study indicates that TRAP-positive monocyte-derived CTGF promotes bone healing by activating PSCs and directing lineage commitment and that targeting PSCs may be an effective strategy for preventing bone non-union.
Collapse
Affiliation(s)
- Yun Bai
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Tao Yu
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jiezhong Deng
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Yusheng Yang
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jiulin Tan
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Qijie Dai
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Zehua Zhang
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Shiwu Dong
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China.,Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, China
| | - Jianzhong Xu
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
20
|
Wei Y, Sun H, Gui T, Yao L, Zhong L, Yu W, Heo SJ, Han L, Dyment NA, Liu XS, Zhang Y, Koyama E, Long F, Zgonis MH, Mauck RL, Ahn J, Qin L. The critical role of Hedgehog-responsive mesenchymal progenitors in meniscus development and injury repair. eLife 2021; 10:e62917. [PMID: 34085927 PMCID: PMC8177886 DOI: 10.7554/elife.62917] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 05/18/2021] [Indexed: 12/18/2022] Open
Abstract
Meniscal tears are associated with a high risk of osteoarthritis but currently have no disease-modifying therapies. Using a Gli1 reporter line, we found that Gli1+ cells contribute to the development of meniscus horns from 2 weeks of age. In adult mice, Gli1+ cells resided at the superficial layer of meniscus and expressed known mesenchymal progenitor markers. In culture, meniscal Gli1+ cells possessed high progenitor activities under the control of Hh signal. Meniscus injury at the anterior horn induced a quick expansion of Gli1-lineage cells. Normally, meniscal tissue healed slowly, leading to cartilage degeneration. Ablation of Gli1+ cells further hindered this repair process. Strikingly, intra-articular injection of Gli1+ meniscal cells or an Hh agonist right after injury accelerated the bridging of the interrupted ends and attenuated signs of osteoarthritis. Taken together, our work identified a novel progenitor population in meniscus and proposes a new treatment for repairing injured meniscus and preventing osteoarthritis.
Collapse
MESH Headings
- Animals
- Cell Lineage
- Cell Proliferation
- Disease Models, Animal
- Hedgehog Proteins/genetics
- Hedgehog Proteins/metabolism
- Humans
- Male
- Menisci, Tibial/metabolism
- Menisci, Tibial/pathology
- Menisci, Tibial/surgery
- Mesenchymal Stem Cell Transplantation
- Mesenchymal Stem Cells/metabolism
- Mice, Knockout
- Osteoarthritis, Knee/genetics
- Osteoarthritis, Knee/metabolism
- Osteoarthritis, Knee/pathology
- Osteoarthritis, Knee/prevention & control
- Signal Transduction
- Swine
- Swine, Miniature
- Tibial Meniscus Injuries/genetics
- Tibial Meniscus Injuries/metabolism
- Tibial Meniscus Injuries/pathology
- Tibial Meniscus Injuries/surgery
- Time Factors
- Wound Healing
- Zinc Finger Protein GLI1/genetics
- Zinc Finger Protein GLI1/metabolism
- Mice
Collapse
Affiliation(s)
- Yulong Wei
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Hao Sun
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen UniversityGuangzhouChina
| | - Tao Gui
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Department of Bone and Joint Surgery, Institute of Orthopedic Diseases, The First Affiliated Hospital, Jinan UniversityGuangzhouChina
| | - Lutian Yao
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Leilei Zhong
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Wei Yu
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Su-Jin Heo
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical CenterPhiladelphiaUnited States
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel UniversityPhiladelphiaUnited States
| | - Nathaniel A Dyment
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Xiaowei Sherry Liu
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Yejia Zhang
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical CenterPhiladelphiaUnited States
- Department of Physical Medicine and Rehabilitation, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Eiki Koyama
- Translational Research Program in Pediatric Orthopaedics, The Children's Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Fanxin Long
- Translational Research Program in Pediatric Orthopaedics, The Children's Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Miltiadis H Zgonis
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Robert L Mauck
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical CenterPhiladelphiaUnited States
| | - Jaimo Ahn
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Department of Orthopaedic Surgery, University of Michigan Medical SchoolAnn ArborUnited States
| | - Ling Qin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
21
|
Yao L, Tichy ED, Zhong L, Mohanty S, Wang L, Ai E, Yang S, Mourkioti F, Qin L. Gli1 Defines a Subset of Fibro-adipogenic Progenitors that Promote Skeletal Muscle Regeneration With Less Fat Accumulation. J Bone Miner Res 2021; 36:1159-1173. [PMID: 33529374 PMCID: PMC8633884 DOI: 10.1002/jbmr.4265] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 01/20/2021] [Accepted: 01/26/2021] [Indexed: 12/21/2022]
Abstract
Skeletal muscle has remarkable regenerative ability after injury. Mesenchymal fibro-adipogenic progenitors (FAPs) are necessary, active participants during this repair process, but the molecular signatures of these cells and their functional relevance remain largely unexplored. Here, using a lineage tracing mouse model (Gli1-CreER Tomato), we demonstrate that Gli1 marks a small subset of muscle-resident FAPs with elevated Hedgehog (Hh) signaling. Upon notexin muscle injury, these cells preferentially and rapidly expanded within FAPs. Ablation of Gli1+ cells using a DTA mouse model drastically reduced fibroblastic colony-forming unit (CFU-F) colonies generated by muscle cells and impaired muscle repair at 28 days. Pharmacologic manipulation revealed that Gli1+ FAPs rely on Hh signaling to increase the size of regenerating myofiber. Sorted Gli1+ FAPs displayed superior clonogenicity and reduced adipogenic differentiation ability in culture compared to sorted Gli1- FAPs. In a glycerol injury model, Gli1+ FAPs were less likely to give rise to muscle adipocytes compared to other FAPs. Further cell ablation and Hh activator/inhibitor treatments demonstrated their dual actions in enhancing myogenesis and reducing adipogenesis after injury. Examining single-cell RNA-sequencing dataset of FAPs from normal mice indicated that Gli1+ FAPs with increased Hh signaling provide trophic signals to myogenic cells while restrict their own adipogenic differentiation. Collectively, our findings identified a subpopulation of FAPs that play an essential role in skeletal muscle repair. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Lutian Yao
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Orthopaedic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Elisia D Tichy
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Leilei Zhong
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Sarthak Mohanty
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Luqiang Wang
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Emily Ai
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Shuying Yang
- Department of Basic & Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA
| | - Foteini Mourkioti
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Penn Institute for Regenerative Medicine, Musculoskeletal Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ling Qin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
22
|
Shuai Y, Lu H, Lv R, Wang J, Wan Q, Mao C, Yang M. Biomineralization Directed by Prenucleated Calcium and Phosphorus Nanoclusters Improving Mechanical Properties and Osteogenic Potential of Antheraea pernyi Silk Fibroin-Based Artificial Periosteum. Adv Healthc Mater 2021; 10:e2001695. [PMID: 33720549 DOI: 10.1002/adhm.202001695] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 01/29/2021] [Indexed: 12/28/2022]
Abstract
The use of biomacromolecules as templates to control the nucleation and growth of hydroxyapatite crystals to prepare bioactive materials is a valuable approach in bone tissue engineering. Here, an artificial periosteum is prepared by biomineralizing Antheraea pernyi fibroin (AF) membrane with prenucleated nanoclusters, which can promote the osteogenic differentiation of mesenchymal stem cells (MSCs) and induce the formation of bone matrix protein in vivo. To achieve this, a biologically inspired prenucleated calcium and phosphorus nanocluster mineralization system is designed to nucleate and generate hydroxyapatite crystals on the surface of the AF membrane. This biomineralization process provides AF membranes with improved elastic modulus and tensile strength. Subsequently, cell viability assay, hemolysis test, and H&E staining show that the mineralized AF (MAF) membranes has good cytocompatibility, hemocompatibility, and histocompatibility in vitro and in vivo. Additionally, the MAF membranes significantly promote osteogenic differentiation of MSCs in the absence of osteogenic inducer in vitro. Experiments in vivo demonstrate that bone-related matrix proteins are highly expressed in MAF groups with or without MSCs seeded. Therefore, the use of bioinspired prenucleated nanoclusters to prepare artificial periosteum based on biomineralized AF membrane is a promising strategy in the field of bone tissue engineering.
Collapse
Affiliation(s)
- Yajun Shuai
- Institute of Applied Bioresource Research College of Animal Science Zhejiang University Hangzhou 310058 China
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province College of Animal Science Zhejiang University Hangzhou 310058 China
| | - Huan Lu
- Institute of Applied Bioresource Research College of Animal Science Zhejiang University Hangzhou 310058 China
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province College of Animal Science Zhejiang University Hangzhou 310058 China
| | - Ruyin Lv
- Institute of Applied Bioresource Research College of Animal Science Zhejiang University Hangzhou 310058 China
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province College of Animal Science Zhejiang University Hangzhou 310058 China
| | - Jie Wang
- Institute of Applied Bioresource Research College of Animal Science Zhejiang University Hangzhou 310058 China
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province College of Animal Science Zhejiang University Hangzhou 310058 China
| | - Quan Wan
- Institute of Applied Bioresource Research College of Animal Science Zhejiang University Hangzhou 310058 China
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province College of Animal Science Zhejiang University Hangzhou 310058 China
| | - Chuanbin Mao
- Department of Chemistry and Biochemistry Stephenson Life Sciences Research Center University of Oklahoma Norman OK 73019‐5300 USA
| | - Mingying Yang
- Institute of Applied Bioresource Research College of Animal Science Zhejiang University Hangzhou 310058 China
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province College of Animal Science Zhejiang University Hangzhou 310058 China
| |
Collapse
|
23
|
Matthews BG, Novak S, Sbrana FV, Funnell JL, Cao Y, Buckels EJ, Grcevic D, Kalajzic I. Heterogeneity of murine periosteum progenitors involved in fracture healing. eLife 2021; 10:e58534. [PMID: 33560227 PMCID: PMC7906599 DOI: 10.7554/elife.58534] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 02/08/2021] [Indexed: 12/15/2022] Open
Abstract
The periosteum is the major source of cells involved in fracture healing. We sought to characterize progenitor cells and their contribution to bone fracture healing. The periosteum is highly enriched with progenitor cells, including Sca1+ cells, fibroblast colony-forming units, and label-retaining cells compared to the endosteum and bone marrow. Using lineage tracing, we demonstrate that alpha smooth muscle actin (αSMA) identifies long-term, slow-cycling, self-renewing osteochondroprogenitors in the adult periosteum that are functionally important for bone formation during fracture healing. In addition, Col2.3CreER-labeled osteoblast cells contribute around 10% of osteoblasts but no chondrocytes in fracture calluses. Most periosteal osteochondroprogenitors following fracture can be targeted by αSMACreER. Previously identified skeletal stem cell populations were common in periosteum but contained high proportions of mature osteoblasts. We have demonstrated that the periosteum is highly enriched with skeletal progenitor cells, and there is heterogeneity in the populations of cells that contribute to mature lineages during periosteal fracture healing.
Collapse
Affiliation(s)
- Brya G Matthews
- Department of Molecular Medicine and Pathology, University of AucklandAucklandNew Zealand
- Department of Reconstructive Sciences, UConn HealthFarmingtonUnited States
| | - Sanja Novak
- Department of Reconstructive Sciences, UConn HealthFarmingtonUnited States
| | - Francesca V Sbrana
- Department of Reconstructive Sciences, UConn HealthFarmingtonUnited States
| | - Jessica L Funnell
- Department of Reconstructive Sciences, UConn HealthFarmingtonUnited States
| | - Ye Cao
- Department of Molecular Medicine and Pathology, University of AucklandAucklandNew Zealand
| | - Emma J Buckels
- Department of Molecular Medicine and Pathology, University of AucklandAucklandNew Zealand
| | - Danka Grcevic
- Department of Physiology and Immunology, University of ZagrebZagrebCroatia
- Croatian Intitute for Brain Research, University of ZagrebZagrebCroatia
| | - Ivo Kalajzic
- Department of Reconstructive Sciences, UConn HealthFarmingtonUnited States
| |
Collapse
|
24
|
Doherty L, Wan M, Kalajzic I, Sanjay A. Diabetes impairs periosteal progenitor regenerative potential. Bone 2021; 143:115764. [PMID: 33221502 PMCID: PMC7770068 DOI: 10.1016/j.bone.2020.115764] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 01/01/2023]
Abstract
Diabetics are at increased risk for fracture, and experience severely impaired skeletal healing characterized by delayed union or nonunion of the bone. The periosteum harbors osteochondral progenitors that can differentiate into chondrocytes and osteoblasts, and this connective tissue layer is required for efficient fracture healing. While bone marrow-derived stromal cells have been studied extensively in the context of diabetic skeletal repair and osteogenesis, the effect of diabetes on the periosteum and its ability to contribute to bone regeneration has not yet been explicitly evaluated. Within this study, we utilized an established murine model of type I diabetes to evaluate periosteal cell differentiation capacity, proliferation, and availability under the effect of a diabetic environment. Periosteal cells from diabetic mice were deficient in osteogenic differentiation ability in vitro, and diabetic mice had reduced periosteal populations of mesenchymal progenitors with a corresponding reduction in proliferation capacity following injury. Additionally, fracture callus mineralization and mature osteoblast activity during periosteum-mediated healing was impaired in diabetic mice compared to controls. We propose that the effect of diabetes on periosteal progenitors and their ability to aid in skeletal repair directly impairs fracture healing.
Collapse
Affiliation(s)
- Laura Doherty
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, UConn Health, Farmington, CT, USA
| | - Matthew Wan
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, UConn Health, Farmington, CT, USA
| | - Ivo Kalajzic
- Department of Reconstructive Sciences, UConn School of Dental Medicine, Farmington, CT, USA
| | - Archana Sanjay
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, UConn Health, Farmington, CT, USA.
| |
Collapse
|
25
|
Kegelman CD, Nijsure MP, Moharrer Y, Pearson HB, Dawahare JH, Jordan KM, Qin L, Boerckel JD. YAP and TAZ Promote Periosteal Osteoblast Precursor Expansion and Differentiation for Fracture Repair. J Bone Miner Res 2021; 36:143-157. [PMID: 32835424 PMCID: PMC7988482 DOI: 10.1002/jbmr.4166] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/10/2020] [Accepted: 07/30/2020] [Indexed: 12/20/2022]
Abstract
In response to bone fracture, periosteal progenitor cells proliferate, expand, and differentiate to form cartilage and bone in the fracture callus. These cellular functions require the coordinated activation of multiple transcriptional programs, and the transcriptional regulators Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) regulate osteochondroprogenitor activation during endochondral bone development. However, recent observations raise important distinctions between the signaling mechanisms used to control bone morphogenesis and repair. Here, we tested the hypothesis that YAP and TAZ regulate osteochondroprogenitor activation during endochondral bone fracture healing in mice. Constitutive YAP and/or TAZ deletion from Osterix-expressing cells impaired both cartilage callus formation and subsequent mineralization. However, this could be explained either by direct defects in osteochondroprogenitor differentiation after fracture or by developmental deficiencies in the progenitor cell pool before fracture. Consistent with the second possibility, we found that developmental YAP/TAZ deletion produced long bones with impaired periosteal thickness and cellularity. Therefore, to remove the contributions of developmental history, we next generated adult onset-inducible knockout mice (using Osx-CretetOff ) in which YAP and TAZ were deleted before fracture but after normal development. Adult onset-induced YAP/TAZ deletion had no effect on cartilaginous callus formation but impaired bone formation at 14 days post-fracture (dpf). Earlier, at 4 dpf, adult onset-induced YAP/TAZ deletion impaired the proliferation and expansion of osteoblast precursor cells located in the shoulder of the callus. Further, activated periosteal cells isolated from this region at 4 dpf exhibited impaired osteogenic differentiation in vitro upon YAP/TAZ deletion. Finally, confirming the effects on osteoblast function in vivo, adult onset-induced YAP/TAZ deletion impaired bone formation in the callus shoulder at 7 dpf before the initiation of endochondral ossification. Together, these data show that YAP and TAZ promote the expansion and differentiation of periosteal osteoblast precursors to accelerate bone fracture healing. © 2020 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Christopher D Kegelman
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Madhura P Nijsure
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Yasaman Moharrer
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Hope B Pearson
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, USA
| | - James H Dawahare
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, USA
| | - Kelsey M Jordan
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Ling Qin
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Joel D Boerckel
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
26
|
Cao Y, Buckels EJ, Matthews BG. Markers for Identification of Postnatal Skeletal Stem Cells In Vivo. Curr Osteoporos Rep 2020; 18:655-665. [PMID: 33034805 DOI: 10.1007/s11914-020-00622-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/29/2020] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW The adult skeleton contains stem cells involved in growth, homeostasis, and healing. Mesenchymal or skeletal stem cells are proposed to provide precursors to osteoblasts, chondrocytes, marrow adipocytes, and stromal cells. We review the evidence for existence and functionality of different skeletal stem cell pools, and the tools available for identifying or targeting these populations in mouse and human tissues. RECENT FINDINGS Lineage tracing and single cell-based techniques in mouse models indicate that multiple pools of stem cells exist in postnatal bone. These include growth plate stem cells, stem and progenitor cells in the diaphysis, reticular cells that only form bone in response to injury, and injury-responsive periosteal stem cells. New staining protocols have also been described for prospective isolation of human skeletal stem cells. Several populations of postnatal skeletal stem and progenitor cells have been identified in mice, and we have an increasing array of tools to target these cells. Most Cre models lack a high degree of specificity to define single populations. Human studies are less advanced and require further efforts to refine methods for identifying stem and progenitor cells in adult bone.
Collapse
Affiliation(s)
- Ye Cao
- Department of Molecular Medicine and Pathology, University of Auckland, Private Bag 92-019, Auckland, 1142, New Zealand
| | - Emma J Buckels
- Department of Molecular Medicine and Pathology, University of Auckland, Private Bag 92-019, Auckland, 1142, New Zealand
| | - Brya G Matthews
- Department of Molecular Medicine and Pathology, University of Auckland, Private Bag 92-019, Auckland, 1142, New Zealand.
| |
Collapse
|
27
|
Xie H, Liu M, Jin Y, Lin H, Zhang Y, Zheng S. miR-1323 suppresses bone mesenchymal stromal cell osteogenesis and fracture healing via inhibiting BMP4/SMAD4 signaling. J Orthop Surg Res 2020; 15:237. [PMID: 32600409 PMCID: PMC7322887 DOI: 10.1186/s13018-020-01685-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/29/2020] [Indexed: 12/14/2022] Open
Abstract
Background Atrophic non-union fractures show no radiological evidence of callus formation within 3 months of fracture. microRNA dysregulation may underlie the dysfunctional osteogenesis in atrophic non-union fractures. Here, we aimed to analyze miR-1323 expression in human atrophic non-union fractures and examine miR-1323’s underlying mechanism of action in human mesenchymal stromal cells. Methods Human atrophic non-union and standard healing fracture specimens were examined using H&E and Alcian Blue staining, immunohistochemistry, qRT-PCR, immunoblotting, and ALP activity assays. The effects of miR-1323 mimics or inhibition on BMP4, SMAD4, osteogenesis-related proteins, ALP activity, and bone mineralization were analyzed in human mesenchymal stromal cells. Luciferase reporter assays were utilized to assay miR-1323’s binding to the 3'UTRs of BMP4 and SMAD4. The effects of miR-1323, BMP4, and SMAD4 were analyzed by siRNA and overexpression vectors. A rat femur fracture model was established to analyze the in vivo effects of antagomiR-1323 treatment. Results miR-1323 was upregulated in human atrophic non-union fractures. Atrophic non-union was associated with downregulation of BMP4 and SMAD4 as well as the osteogenic markers ALP, collagen I, and RUNX2. In vitro, miR-1323 suppressed BMP4 and SMAD4 expression by binding to the 3'UTRs of BMP4 and SMAD4. Moreover, miR-1323’s inhibition of BMP4 and SMAD4 inhibited mesenchymal stromal cell osteogenic differentiation via modulating the nuclear translocation of the transcriptional co-activator TAZ. In vivo, antagomiR-1323 therapy facilitated the healing of fractures in a rat model of femoral fracture. Conclusions This evidence supports the miR-1323/BMP4 and miR-1323/SMAD4 axes as novel therapeutic targets for atrophic non-union fractures.
Collapse
Affiliation(s)
- Hui Xie
- Department of Orthopedics, The Second Affiliated Hospital of Jiaxing University, No. 1518 Huanchengbei Road, Jiaxing, Zhejiang, 314299, China
| | - Ming Liu
- Department of Orthopedics, The Second Affiliated Hospital of Jiaxing University, No. 1518 Huanchengbei Road, Jiaxing, Zhejiang, 314299, China
| | - Yaofeng Jin
- Department of Orthopedics, The Second Affiliated Hospital of Jiaxing University, No. 1518 Huanchengbei Road, Jiaxing, Zhejiang, 314299, China
| | - Haiqing Lin
- Department of Orthopedics, The Second Affiliated Hospital of Jiaxing University, No. 1518 Huanchengbei Road, Jiaxing, Zhejiang, 314299, China
| | - Yushan Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Jiaxing University, No. 1518 Huanchengbei Road, Jiaxing, Zhejiang, 314299, China
| | - Song Zheng
- Department of Orthopedics, The Second Affiliated Hospital of Jiaxing University, No. 1518 Huanchengbei Road, Jiaxing, Zhejiang, 314299, China.
| |
Collapse
|
28
|
Kaur A, Mohan S, Rundle CH. A segmental defect adaptation of the mouse closed femur fracture model for the analysis of severely impaired bone healing. Animal Model Exp Med 2020; 3:130-139. [PMID: 32613172 PMCID: PMC7323699 DOI: 10.1002/ame2.12114] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 03/16/2020] [Accepted: 04/02/2020] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVE To better characterize nonunion endochondral bone healing and evaluate novel therapeutic approaches for critical size defect healing in clinically challenging bone repair, a segmental defect model of bone injury was adapted from the three-point bending closed fracture technique in the murine femur. METHODS The mouse femur was surgically stabilized with an intramedullary threaded rod with plastic spacers and the defect adjusted to different sizes. Healing of the different defects was analyzed by radiology and histology to 8 weeks postsurgery. To determine whether this model was effective for evaluating the benefits of molecular therapy, BMP-2 was applied to the defect and healing then examined. RESULTS Intramedullary spacers were effective in maintaining the defect. Callus bone formation was initiated but was arrested at defect sizes of 2.5 mm and above, with no more progress in callus bone development evident to 8 weeks healing. Cartilage development in a critical size defect attenuated very early in healing without bone development, in contrast to the closed femur fracture healing, where callus cartilage was replaced by bone. BMP-2 therapy promoted osteogenesis of the resident cells of the defect, but there was no further callus development to indicate that healing to pre-surgery bone structure was successful. CONCLUSIONS This segmental defect adaptation of the closed femur fracture model of murine bone repair severely impairs callus development and bone healing, reflecting a challenging bone injury. It is adjustable and can be compared to the closed fracture model to ascertain healing deficiencies and the efficacy of therapeutic approaches.
Collapse
Affiliation(s)
- Amandeep Kaur
- Musculoskeletal Disease CenterResearch Service (151)Jerry L. Pettis Memorial Veterans Administration Medical CenterLoma LindaCAUSA
| | - Subburaman Mohan
- Musculoskeletal Disease CenterResearch Service (151)Jerry L. Pettis Memorial Veterans Administration Medical CenterLoma LindaCAUSA
- Department of MedicineLoma Linda UniversityLoma LindaCAUSA
- Department of Orthopedic SurgeryLoma Linda UniversityLoma LindaCAUSA
| | - Charles H. Rundle
- Musculoskeletal Disease CenterResearch Service (151)Jerry L. Pettis Memorial Veterans Administration Medical CenterLoma LindaCAUSA
- Department of MedicineLoma Linda UniversityLoma LindaCAUSA
| |
Collapse
|
29
|
Liu Y, Fang J, Zhang Q, Zhang X, Cao Y, Chen W, Shao Z, Yang S, Wu D, Hung M, Zhang Y, Tong W, Tian H. Wnt10b-overexpressing umbilical cord mesenchymal stem cells promote critical size rat calvarial defect healing by enhanced osteogenesis and VEGF-mediated angiogenesis. J Orthop Translat 2020; 23:29-37. [PMID: 32477867 PMCID: PMC7248289 DOI: 10.1016/j.jot.2020.02.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 02/08/2020] [Accepted: 02/13/2020] [Indexed: 01/04/2023] Open
Abstract
Background/objectives Accelerating the process of bone regeneration is of great interest for surgeons and basic scientists alike. Recently, umbilical cord mesenchymal stem cells (UCMSCs) are considered clinically applicable for tissue regeneration due to their noninvasive harvesting and better viability. Nonetheless, the bone regenerative ability of human UCMSCs (HUCMSCs) is largely unknown. This study aimed to investigate whether Wnt10b-overexpressing HUCMSCs have enhanced bone regeneration ability in a rat model. Method A rat calvarial defect was performed on 8-week old male Sprague Dawley rats. Commercially purchased HUCMSCsEmp in hydrogel, HUCMSCsWnt10b in hydrogel and HUCMSCsWnt10b with IWR-1 were placed in the calvarial bone defect right after surgery on rats (N = 8 rats for each group). Calvaria were harvested for micro-CT analysis and histology four weeks after surgery. CFU-F and multi-differentiation assay by oil red staining, alizarin red staining and RT-PCR (real-time polymerase chain reaction) were performed on HUCMSCsEmp and HUCMSCsWnt10bin vitro. Conditioned media from HUCMSCsEmp and HUCMSCsWnt10b were collected and used to treat human umbilical cord vein endothelial cells in Matrigel to access vessel formation capacity by tube formation assay. Results Alizarin red staining, oil red staining and RT-PCR results showed robust osteogenic differentiation but poor adipogenic differentiation ability of HUCMSCsWnt10b. Furthermore, HUCMSCsWnt10b could accelerate bone defect healing, which was likely due to enhanced angiogenesis after the HUCMSCsWnt10b treatment, because more CD31+ vessels and increased vascular endothelial growth factor-A (VEGF-A) expression were observed, compared with the HUCMSCsEmp treatment. Conditioned media from HUCMSCsWnt10b also induced endothelial cells to form vessel tubes in a tube formation assay, which could be abolished by SU5416, an angiogenesis inhibitor. Conclusion To our knowledge, this is the first study providing empirical evidence that HUCMSCsWnt10b can enhance their ability to heal calvarial bone defects via VEGF-mediated angiogenesis. The translational potential of this article HUCMSCsWnt10b can accelerate critical size calvaria and are a new promising therapeutic cell source for fracture nonunion healing.
Collapse
Affiliation(s)
- Yong Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277, Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Jiarui Fang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277, Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Quan Zhang
- Wuhan Hamilton Biotechnology Co., Ltd, Wuhan, Hubei, 430075, China
| | - Xiaoguang Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277, Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Yulin Cao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277, Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Wei Chen
- The Third Hospital of Hebei Medical University, 139, Ziqiang Road, Shi Jiazhuang, Hebei, 050051, China
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277, Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Shuhua Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277, Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Dongcheng Wu
- Wuhan Hamilton Biotechnology Co., Ltd, Wuhan, Hubei, 430075, China.,Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Wuhan University, China
| | - Man Hung
- College of Dental Medicine, Roseman University of Health Sciences, 10984 S River Front Pkwy, South Jordan, UT, 84095, USA
| | - Yingze Zhang
- The Third Hospital of Hebei Medical University, 139, Ziqiang Road, Shi Jiazhuang, Hebei, 050051, China
| | - Wei Tong
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277, Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Hongtao Tian
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277, Jiefang Avenue, Wuhan, Hubei, 430022, China
| |
Collapse
|